Knock-In of an Internal Tandem Duplication Mutation into Murine FLT3 Confers Myeloproliferative Disease in a Mouse Model.

Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 386-386
Author(s):  
Li Li ◽  
Obdulio Piloto ◽  
Ho Bao Nguyen ◽  
Kathleen Greenberg ◽  
Frederick Racke ◽  
...  

Abstract Activation of FLT3 by internal tandem duplication (ITD) mutations in the juxtamembrane domain is the most common molecular alteration known in AML and confers poor prognosis. Homozygous or hemizygous ITD mutations, with the loss of wild type FLT3, result in even worse prognosis. It has been previously reported that FLT3/ITD activates different signaling pathways as compared to wt FLT3. Several models designed to study the roles of FLT3/ITD in leukemogenesis have been reported, utilizing either retroviral infection of murine BM or transgenic expression off strong promoters. Those models have shown myeloproliferative disease (MPD) with FLT3/ITD expression alone and full transformation to leukemia when additional genetic alterations are added. However, these models utilize high level expression of FLT3/ITD, and potentially express the gene at inappropriate stages of development. In addition, retroviral integration sites could be playing an active role. To more closely simulate and study the in vivo biological impact of FLT3/ITD mutations in the development of leukemia, we generated a FLT3/ITD “knock-in” mouse model by inserting an ITD mutation into the juxtamembrane region of murine FLT3 genomic DNA. Young FLT3 wt/ITD mice showed signs of MPD which progressed to fatality at the age of 6–16 months. Older FLT3wt/ITD mice had significant splenomegaly with disruption of the normal splenic architecture. Increased WBC and reduced RBC counts were detected in the peripheral blood of these mice, with elevated monocyte and neutrophil counts. BM was hypercellular with an increased fraction of granulocytes/monocytes and their progenitors, dendritic cells (DCs), and a reduction in the fraction of B lymphocytes. No signs of leukemia were observed over the lifetime of these mice. Clonogenic assay demonstrated that BM from FLT3wt/ITD mice contained increased granulocytic/monocytic colony-forming cells. In addition, these cells were immortalized as they could undergo long-term serial culture in cytokine-supplemented liquid medium. BM from FLT3wt/ITD mice also generated more colonies in the in vivo spleen-CFU assay, and showed an enhanced ability to repopulate lethally irradiated recipients in the long-term competitive repopulation assay. FLT3ITD/ITD mice developed fatal MPD with a shorter latency. Leukocytosis and BM hypercellularity were more pronounced for these mice, with an even higher fraction of granulocytic/monocytic progenitors and a tremendous suppression of B lymphocytes in the BM. BM from these mice displayed reduced potential for reopulation in the long-term competitive repopulation assay. Some FLT3ITD/ITD mice spontaneously developed leukemia-like disease with a lag of 2–6 months. Histopathology revealed complete effacement of the normal spleen architecture, extensive infiltration of the liver and meninges, and complete replacement of the BM with immature myeloid cells. In summary, these data indicate that expression of a FLT3/ITD mutation alone is capable of partially transforming normal hematopoietic stem cells and progenitors to a phenotype of MPD. Additional cooperative events are likely required to progress to leukemia. These will serve as excellent models to study the pathways by which FLT3/ITD signaling contributes to leukemogenesis.

Blood ◽  
2008 ◽  
Vol 111 (7) ◽  
pp. 3849-3858 ◽  
Author(s):  
Li Li ◽  
Obdulio Piloto ◽  
Ho Bao Nguyen ◽  
Kathleen Greenberg ◽  
Kogo Takamiya ◽  
...  

Abstract Constitutive activation of FMS-like tyrosine kinase 3 (FLT3) by internal tandem duplication (ITD) mutations is one of the most common molecular alterations known in acute myeloid leukemia (AML). To investigate the role FLT3/ITD mutations play in the development of leukemia, we generated a FLT3/ITD knock-in mouse model by inserting an ITD mutation into the juxtamembrane domain of murine Flt3. FLT3wt/ITD mice developed myeloproliferative disease, characterized by splenomegaly, leukocytosis, and myeloid hypercellularity, which progressed to mortality by 6 to 20 months. Bone marrow (BM) and spleen from FLT3wt/ITD mice had an increased fraction of granulocytes/monocytes and dendritic cells, and a decreased fraction of B-lymphocytes. No sign of acute leukemia was observed over the lifetime of these mice. BM from FLT3wt/ITD mice showed enhanced potential to generate myeloid colonies in vitro. BM from FLT3wt/ITD mice also produced more spleen colonies in the in vivo colony-forming unit (CFU)–spleen assay. In the long-term competitive repopulation assay, BM cells from FLT3wt/ITD mice outgrew the wild-type competitor cells and showed increased myeloid and reduced lymphoid expansion activity. In summary, our data indicate that expression of FLT3/ITD mutations alone is capable of conferring normal hematopoietic stem/progenitor cells (HSPCs) with enhanced myeloid expansion. It also appears to suppress B lymphoid maturation. Additional cooperative events appear to be required to progress to acute leukemia.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 1356-1356
Author(s):  
Christian Brandts ◽  
Miriam Rode ◽  
Beate Lindtner ◽  
Gabriele Koehler ◽  
Steffen Koschmieder ◽  
...  

Abstract Activating mutations in Flt3, N- and K-Ras have been reported in all AML subtypes and represent common molecular defects in de novo AML. We have previously shown that these mutations lead to constitutive AKT phosphorylation and activation. As a consequence, Akt phosphorylation is found in myeloid blasts of the majority of AML patients. We reasoned that constitutively active AKT may contribute to leukemia development, and therefore we assessed the contribution of AKT in oncogenic transformation in vivo. For this purpose, we established an inducible mouse model expressing myristylated AKT1 under the control of the scl-3′ enhancer (MyrAKT1). This system restricts activated AKT1 to endothelium, hematopoietic stem cells and myeloid lineage cells at a low but detectable level. About 40% of induced mice developed a myeloproliferative disorder after latencies of 7 to 22 months. Onset of disease was frequently associated with hemangioma formation, due to endothelial MyrAKT1 expression. The myeloproliferative disorder was associated with splenomegaly with increased extramedullary hematopoiesis, while the peripheral blood contained mature granulocytes. Furthermore, the stem cell and progenitor cell compartment in spleens and bone marrow of these mice was altered compared to control mice. Colony formation assays with MyrAKT1-expressing bone marrow suggested that overactivation of AKT1 enhanced proliferation. The AKT1-induced disease was transplantable by both bone marrow and spleen cells. These findings highlight the oncogenic capacity of constitutively activated AKT1 in vivo and indicate that AKT is an attractive target for therapeutic intervention in AML.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 1420-1420
Author(s):  
S. Haihua Chu ◽  
Diane Heiser ◽  
Li Li ◽  
Ian M Kaplan ◽  
Curt I. Civin ◽  
...  

Abstract Abstract 1420 Poster Board I-443 As one of the most common genetic alterations found in acute myeloid leukemia (AML), constitutive activation of the FMS-like tyrosine kinase (FLT3) has provided a promising candidate for small molecule targeted therapy. However, the results of FLT3 inhibitor monotherapy trials indicate FLT3 inhibition alone is insufficient to induce consistent and durable responses. Moreover, after an initial response, many patients relapse, suggesting that leukemia-initiating stem cells may be escaping inhibitor-induced cytotoxicity. Currently, the exact stage at which activating mutations in FLT3 occur during transformation is unknown. While FLT3 knockout mice have minor defects in hematopoiesis, very little is known about either the effect of FLT3 activating mutations on normal hematopoietic stem cells or the contribution of FLT3 activation to leukemogenesis. Thus, in order to better understand the underlying molecular mechanisms of transformation and to identify novel targets for treatment of AML, the role of FLT3 activating mutations in hematopoietic stem cells (HSCs) is of great interest. To study the natural stem cell reservoir and other populations that may escape inhibition, our laboratory has developed a knock-in mouse model in which the FLT3/ITD mutation (an internal tandem duplication correlated with poor prognosis in patients) has been introduced under the endogenous promoter, resulting in myeloproliferative disease (MPD). Conventional transplantation using unfractionated or lineage-depleted marrow from FLT3/ITD mice failed to fully engraft or recapitulate disease, suggesting a HSC defect. Thus, in order to identify a compartment enriched for MPD-initiating cells, several cell surface marker-defined hematopoietic populations were transplanted and compared for engraftment and disease recapitulation. Lineage negative (LIN-), KSL (KIT+SCA+LIN-), MPP (KSL CD34+FLT3+), and ST-HSC (KSL CD34+FLT3-) cells sorted from FLT3/ITD bone marrow all had significantly reduced reconstitution capacity compared to the same compartment from WT littermates. In contrast, highly purified LT-HSCs (KSL CD34-FLT3-) generated equivalent engraftment whether from WT or ITD bone marrow. Furthermore, we measured Hoechst dye efflux in WT and ITD bone marrow to examine side population (SP) cells, known to be enriched in HSC activity, and found that FLT3/ITD mice displayed five-fold fewer SP cells. In addition, bone marrow from FLT3/ITD mice showed a ten-fold decrease in SLAM-defined stem cell frequency (LIN-CD48-CD41-CD150+). 500 sorted SLAM cells from either WT or FLT3/ITD mice were sufficient to fully reconstitute a transplant recipient, demonstrating an equivalent engraftment capacity within this HSC-enriched compartment. Moreover, the MPD phenotype was successfully recapitulated in primary transplants of FLT3/ITD SLAM cells as characterized by an increase in myeloid progenitors, expansion of the LIN- fraction, enlarged spleens and depletion of the SLAM compartment compared to WT SLAM transplant recipients. Classically defined as a class II oncogene, FLT3 activating mutations have been shown to drive proliferation in cells harboring the mutation. To investigate whether FLT3/ITD drives proliferation in the most primitive hematopoietic compartments, BrdU incorporation was examined in FLT3/ITD hematopoietic stem and progenitor-enriched subsets. While myeloid progenitor compartments showed a decrease in proliferation as compared to WT littermates, the FLT3/ITD SLAM and KSL compartments had an increased percentage of BrdU-incorporating cells. Altogether, our data suggests a role for FLT3/ITD in driving normally quiescent HSCs to proliferate, thereby depleting the pool of primitive HSCs. In this model, HSC depletion coupled to rapid expansion in progenitor cell numbers leads to perturbation of normal hematopoiesis giving rise to a myeloproliferative disease. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
1997 ◽  
Vol 90 (4) ◽  
pp. 1701-1708 ◽  
Author(s):  
Robert G. Andrews ◽  
Aaron Winkler ◽  
Jennifer Potter ◽  
Eileen Bryant ◽  
Glenn H. Knitter ◽  
...  

Abstract The CD34 antigen is thought to be expressed by hematopoietic stem cells in adult humans and nonhuman primates. We present data that baboons transplanted with highly purified allogeneic CD34+ marrow cells devoid of detectable mature and immature T and B lymphocytes and myeloid cells, isolated from sex-mismatched mixed lymphocyte culture (MLC) nonreactive siblings, have maintained stable lymphohematopoietic engraftment with donor cells for greater than 4.9, greater than 6.0, and 5.0 years. Cytogenetic analysis of unfractionated marrow and peripheral blood cells at multiple time points after transplantation show virtually all donor cells in two animals and stable mixed chimerism in the third. We used polymerase chain reaction to show that colony-forming unit–granulocyte-macrophage, burst-forming unit-erythroid, and high proliferative potential colony-forming cells (HPP-CFC) were virtually all of donor origin in two animals and present at lower levels in the stable mixed chimera. CD20+ B-lymphoblastoid cell lines derived by Herpesvirus Papio transformation of peripheral blood cells were virtually all donor in two animals and 50% donor in the mixed chimera. CD4+ and CD8+ T cells and neutrophils purified from the peripheral blood of the two female animals also were all donor-derived. To assess immunologic function after transplantation, we immunized the three long-term chimeric animals and two normal control animals with bacteriophage ΦX-174, a neoantigen that requires the interaction of antigen-presenting cells, T lymphocytes, and B lymphocytes to mount a normal antibody response. Experimental and control animals, when immunized with bacteriophage, had similar serum Ig levels. The experimental and control animals generated similar titers of antibacteriophage antibodies after primary and secondary immunizations with evidence of amplification and class switching. These findings further support the hypothesis that the CD34+ antigen is expressed on hematopoietic stem cells that can mediate stable long-term lymphohematopoiesis in vivo and, importantly, that normal immunologic function can be reconstituted in vivo after transplantation of the highly purified CD34+ Lin− cells alone.


Blood ◽  
1997 ◽  
Vol 90 (4) ◽  
pp. 1701-1708
Author(s):  
Robert G. Andrews ◽  
Aaron Winkler ◽  
Jennifer Potter ◽  
Eileen Bryant ◽  
Glenn H. Knitter ◽  
...  

The CD34 antigen is thought to be expressed by hematopoietic stem cells in adult humans and nonhuman primates. We present data that baboons transplanted with highly purified allogeneic CD34+ marrow cells devoid of detectable mature and immature T and B lymphocytes and myeloid cells, isolated from sex-mismatched mixed lymphocyte culture (MLC) nonreactive siblings, have maintained stable lymphohematopoietic engraftment with donor cells for greater than 4.9, greater than 6.0, and 5.0 years. Cytogenetic analysis of unfractionated marrow and peripheral blood cells at multiple time points after transplantation show virtually all donor cells in two animals and stable mixed chimerism in the third. We used polymerase chain reaction to show that colony-forming unit–granulocyte-macrophage, burst-forming unit-erythroid, and high proliferative potential colony-forming cells (HPP-CFC) were virtually all of donor origin in two animals and present at lower levels in the stable mixed chimera. CD20+ B-lymphoblastoid cell lines derived by Herpesvirus Papio transformation of peripheral blood cells were virtually all donor in two animals and 50% donor in the mixed chimera. CD4+ and CD8+ T cells and neutrophils purified from the peripheral blood of the two female animals also were all donor-derived. To assess immunologic function after transplantation, we immunized the three long-term chimeric animals and two normal control animals with bacteriophage ΦX-174, a neoantigen that requires the interaction of antigen-presenting cells, T lymphocytes, and B lymphocytes to mount a normal antibody response. Experimental and control animals, when immunized with bacteriophage, had similar serum Ig levels. The experimental and control animals generated similar titers of antibacteriophage antibodies after primary and secondary immunizations with evidence of amplification and class switching. These findings further support the hypothesis that the CD34+ antigen is expressed on hematopoietic stem cells that can mediate stable long-term lymphohematopoiesis in vivo and, importantly, that normal immunologic function can be reconstituted in vivo after transplantation of the highly purified CD34+ Lin− cells alone.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 865-865
Author(s):  
Sarah M Greenblatt ◽  
Li Li ◽  
Christopher Slape ◽  
Rachel L Novak ◽  
Amy S. Duffield ◽  
...  

Abstract Abstract 865 Acute myeloid leukemia (AML) with myelodysplastic related changes represents 24–35% of all cases of AML and has a poor response to chemotherapy and a dismal prognosis. Activating mutations of the FMS-like tyrosine kinase 3 (FLT3) receptor have been seen in 5% of myelodysplastic syndrome (MDS) cases, and an additional 10% of patients with MDS acquire FLT3 mutations during progression to AML. We have previously generated a knock-in mouse model in which an internal tandem duplication (ITD) mutation was inserted into the murine Flt3 gene, which induces a lethal myeloproliferative neoplasm, but not progression to overt leukemia. One mouse model of MDS involves the transgenic expression of a Nup98-HoxD13 (NHD13) fusion under the hematopoietic specific vav promoter. We bred the FLT3/ITD knock-in mice to the NHD13 transgenic mice to see if the two genetic alterations would cooperate. Strikingly, the FLT3/ITD-NHD13 mice on the FVB/N background developed acute leukemia with 100% penetrance and a mean survival of 95 ± 32 days (p<0.0001, n=20). This compares with a mean survival of 281 ± 94 days and 372 ± 84 days for the NHD13 alone and FLT3/ITD alone mice, respectively (both p<0.0001, n=20). FLT3/ITD-NHD13 mice generated on the C57Bl/6N background developed leukemia with a longer latency of 143 ± 37 days, but they still had a significantly shorter survival compared to the single mutants alone. Competitive repopulation experiments showed that leukemic bone marrow was able to engraft in lethally irradiated recipients, with 1:200 cells in the bulk bone marrow having the potential to generate leukemia in recipient mice. To determine the identity of the leukemic initiating cell (LIC), the bulk bone marrow was further sorted into the multipotent progenitor (MPP), common myeloid progenitor (CMP), granulocyte/macrophage progenitor (GMP), and megakaryocytic/erythroid progenitor (MEP) populations and limiting dilution transplantation was performed for each group. The highest engraftment potential was found in the MPP population (1:100 cells) with much rarer LICs from the CMP and GMP. The MEP population did not engraft in any recipient mice even at the highest cell dose used of 100,000 sorted cells. RNA was extracted from the total bone marrow of mice 3 months prior to the development of disease and showed overexpression of HoxA7, HoxA9, HoxB4, HoxB6, HoxC4, and HoxC6 in the FLT3/ITD-NHD13 mice compared to the age-matched wildtype or FLT3/ITD mice. The overexpression of these genes appeared to be primarily driven by the NHD13 transgene, although there was also an increase in HoxA7 and HoxA9 expression observed in the FLT3/ITD mice. Since Hox genes are known to play an important role in stem cell self-renewal, we isolated RNA from 2-month-old littermates and stained for the cell surface markers characterizing the KSL, MPP, ST-HSC, and LT-HSC populations. FLT3/ITD-NHD13 and FLT3/ITD mice had a significant increase in the percentage of KSLs, long term HSCs, and short term HSCs. FLT3/ITD mice showed increased numbers of MPPs with a smaller but significant increase in the FLT3/ITD-NHD13 cohort. A significant number of AML patients with FLT3/ITD mutations present with loss of the wildtype allele of FLT3 and additional patients acquire this loss at the time of relapse. Hemizygosity at the FLT3 locus in FLT3/ITD mutant AML patients is associated with an even more adverse prognosis compared to patients with an intact wild-type allele. We observed spontaneous loss of heterozygosity (LOH) occurring in 100% of the FLT3/ITD-NHD13 mice and resulting in loss of the wildtype Flt3 allele. The frequent LOH seen in the FLT3/ITD-NHD13 mice provides additional evidence that this transgenic mouse serves as an accurate model to further explore the molecular pathways that underlie the development of leukemias with activating FLT3 mutations. This mouse model is also one of the first models of MDS-related AML and should provide a means to explore the molecular pathways that underlie these devastating hematopoietic neoplasms. Disclosures: Borowitz: BD Biosciences: Research Funding.


Author(s):  
Fatima Aerts-Kaya

: In contrast to their almost unlimited potential for expansion in vivo and despite years of dedicated research and optimization of expansion protocols, the expansion of Hematopoietic Stem Cells (HSCs) in vitro remains remarkably limited. Increased understanding of the mechanisms that are involved in maintenance, expansion and differentiation of HSCs will enable the development of better protocols for expansion of HSCs. This will allow procurement of HSCs with long-term engraftment potential and a better understanding of the effects of the external influences in and on the hematopoietic niche that may affect HSC function. During collection and culture of HSCs, the cells are exposed to suboptimal conditions that may induce different levels of stress and ultimately affect their self-renewal, differentiation and long-term engraftment potential. Some of these stress factors include normoxia, oxidative stress, extra-physiologic oxygen shock/stress (EPHOSS), endoplasmic reticulum (ER) stress, replicative stress, and stress related to DNA damage. Coping with these stress factors may help reduce the negative effects of cell culture on HSC potential, provide a better understanding of the true impact of certain treatments in the absence of confounding stress factors. This may facilitate the development of better ex vivo expansion protocols of HSCs with long-term engraftment potential without induction of stem cell exhaustion by cellular senescence or loss of cell viability. This review summarizes some of available strategies that may be used to protect HSCs from culture-induced stress conditions.


Blood ◽  
1996 ◽  
Vol 87 (10) ◽  
pp. 4136-4142 ◽  
Author(s):  
I Kawashima ◽  
ED Zanjani ◽  
G Almaida-Porada ◽  
AW Flake ◽  
H Zeng ◽  
...  

Using in utero transplantation into fetal sheep, we examined the capability of human bone marrow CD34+ cells fractionated based on Kit protein expression to provide long-term in vivo engraftment. Twelve hundred to 5,000 CD34+ Kit-, CD34+ Kit(low), and CD34+ Kit(high) cells were injected into a total of 14 preimmune fetal sheep recipients using the amniotic bubble technique. Six fetuses were killed in utero 1.5 months after bone marrow cell transplantation. Two fetuses receiving CD34+ Kit(low) cells showed signs of engraftment according to analysis of CD45+ cells in their bone marrow cells and karyotype studies of the colonies grown in methylcellulose culture. In contrast, two fetuses receiving CD34+ Kit(high) cells and two fetuses receiving CD34+ Kit- cells failed to show evidence of significant engraftment. Two fetuses were absorbed. A total of six fetuses receiving different cell populations were allowed to proceed to term, and the newborn sheep were serially examined for the presence of chimerism. Again, only the two sheep receiving CD34+ Kit(low) cells exhibited signs of engraftment upon serial examination. Earlier in studies of murine hematopoiesis, we have shown stage-specific changes in Kit expression by the progenitors. The studies of human cells reported here are in agreement with observations in mice, and indicate that human hematopoietic stem cells are enriched in the Kit(low) population.


2012 ◽  
Vol 45 ◽  
pp. S89 ◽  
Author(s):  
Floor M. Lambers ◽  
Kathleen Koch ◽  
Gisela Kuhn ◽  
Claudia Weigt ◽  
Friederike A. Schulte ◽  
...  
Keyword(s):  

Blood ◽  
1989 ◽  
Vol 74 (3) ◽  
pp. 930-939 ◽  
Author(s):  
SJ Szilvassy ◽  
PM Lansdorp ◽  
RK Humphries ◽  
AC Eaves ◽  
CJ Eaves

Abstract A simple procedure is described for the quantitation and enrichment of murine hematopoietic cells with the capacity for long-term repopulation of lymphoid and myeloid tissues in lethally irradiated mice. To ensure detection of the most primitive marrow cells with this potential, we used a competitive assay in which female recipients were injected with male “test” cells and 1 to 2 x 10(5) “compromised” female marrow cells with normal short-term repopulating ability, but whose long-term repopulating ability had been reduced by serial transplantation. Primitive hematopoietic cells were purified by flow cytometry and sorting based on their forward and orthogonal light-scattering properties, and Thy-1 and H-2K antigen expression. Enrichment profiles for normal marrow, and marrow of mice injected with 5-fluorouracil (5- FU) four days previously, were established for each of these parameters using an in vitro assay for high proliferative potential, pluripotent colony-forming cells. When all four parameters were gated simultaneously, these clonogenic cells were enriched 100-fold. Both day 9 and day 12 CFU-S were copurified; however, the purity (23%) and enrichment (75-fold) of day 12 CFU-S in the sorted population was greater with 5-FU-treated cells. Five hundred of the sorted 5-FU marrow cells consistently repopulated recipient lymphoid and myeloid tissues (greater than 50% male, 1 to 3 months post-transplant) when co-injected with 1 to 2 x 10(5) compromised female marrow cells, and approximately 100 were sufficient to achieve the same result in 50% of recipients under the same conditions. This relatively simple purification and assay strategy should facilitate further analysis of the heterogeneity and regulation of stem cells that maintain hematopoiesis in vivo.


Sign in / Sign up

Export Citation Format

Share Document